Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
SLAS Technol ; 23(4): 338-351, 2018 08.
Article in English | MEDLINE | ID: mdl-29361868

ABSTRACT

Reliable cell recovery and expansion are fundamental to the successful scale-up of chimeric antigen receptor (CAR) T cells or any therapeutic cell-manufacturing process. Here, we extend our previous work in whole blood by manufacturing a highly parallel deterministic lateral displacement (DLD) device incorporating diamond microposts and moving into processing, for the first time, apheresis blood products. This study demonstrates key metrics of cell recovery (80%) and platelet depletion (87%), and it shows that DLD T-cell preparations have high conversion to the T-central memory phenotype and expand well in culture, resulting in twofold greater central memory cells compared to Ficoll-Hypaque (Ficoll) and direct magnetic approaches. In addition, all samples processed by DLD converted to a majority T-central memory phenotype and did so with less variation, in stark contrast to Ficoll and direct magnetic prepared samples, which had partial conversion among all donors (<50%). This initial comparison of T-cell function infers that cells prepared via DLD may have a desirable bias, generating significant potential benefits for downstream cell processing. DLD processing provides a path to develop a simple closed system that can be automated while simultaneously addressing multiple steps when there is potential for human error, microbial contamination, and other current technical challenges associated with the manufacture of therapeutic cells.


Subject(s)
Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Blood Component Removal , Cell Proliferation , Cell Separation , Humans , Lymphocyte Activation/immunology , Microarray Analysis , Phenotype
2.
Methods Mol Biol ; 717: 3-43, 2011.
Article in English | MEDLINE | ID: mdl-21370022

ABSTRACT

Protein phosphorylation is a universal key posttranslational modification that affects the activity and other properties of intracellular proteins. Phosphosite-specific antibodies can be produced as polyclonals or monoclonals in different animal species, and each approach offers its own benefits and disadvantages. The validation of phosphosite-specific antibodies requires multiple techniques and tactics to demonstrate their specificity. These antibodies can be used in arrays, flow cytometry, and imaging platforms. The specificity of phosphosite-specific antibodies is key for their use in proteomics and profiling of disease.


Subject(s)
Antibodies, Phospho-Specific/analysis , Immunohistochemistry/methods , Proteins/analysis , Animals , Antibodies, Phospho-Specific/immunology , Blotting, Western/methods , Drug Discovery/methods , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Humans , Phosphorylation , Protein Processing, Post-Translational , Proteins/immunology , Proteins/metabolism , Proteomics/methods , Validation Studies as Topic
3.
Blood ; 112(13): 5254-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18838616

ABSTRACT

Graft-versus-host disease (GVHD) is a serious complication of allogeneic bone marrow transplantation, and donor T cells are indispensable for GVHD. Current therapies have limited efficacy, selectivity, and high toxicities. We used a novel flow cytometry technique for the analysis of intracellular phosphorylation events in single cells in murine BMT models to identify and validate novel GVHD drug targets.(1-7) This method circumvents the requirement for large numbers of purified cells, unlike western blots. We defined a signaling profile for alloactivated T cells in vivo and identified the phosphorylation of ERK1/2 and STAT-3 as important events during T-cell (allo)activation in GVHD. We establish that interference with STAT-3 phosphorylation can inhibit T-cell activation and proliferation in vitro and GVHD in vivo. This suggests that phospho-specific flow cytometry is useful for the identification of promising drug targets, and ERK1/2 and STAT-3 phosphorylation in alloactivated T cells may be important for GVHD.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease , Lymphocyte Activation , Mitogen-Activated Protein Kinase 3/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Animals , Flow Cytometry , Mice , Phosphorylation/immunology , Transplantation, Homologous
4.
Mol Cell Biol ; 28(10): 3489-501, 2008 May.
Article in English | MEDLINE | ID: mdl-18347059

ABSTRACT

The target of rapamycin (TOR) signaling regulates the nucleocytoplasmic shuttling of transcription factors in yeast. Whether the mammalian counterpart of TOR (mTOR) also regulates nucleocytoplasmic shuttling is not known. Using a phospho-specific monoclonal antibody, we demonstrate that mTOR phosphorylates Ser(168,170) of endogenous NFATc4, which are conserved gate-keeping Ser residues that control NFAT subcellular distribution. The mTOR acts as a basal kinase during the resting state to maintain NFATc4 in the cytosol. Inactivation and nuclear export of NFATc4 are mediated by rephosphorylation of Ser(168,170), which can be a nuclear event. Kinetic analyses demonstrate that rephosphorylation of Ser(168,170) of endogenous NFATc4 is mediated by mTOR and, surprisingly, by extracellular signal-regulated kinase 5 (ERK5) mitogen-activated protein kinase as well. Ablation of ERK5 in the Erk5(-/-) cells ascertains defects in NFATc4 rephosphorylation and nucleocytoplasmic shuttling. In addition, phosphorylation of NFATc4 by ERK5 primes subsequent phosphorylation mediated by CK1alpha. These results demonstrate that distinct protein kinases are integrated to phosphorylate the gate-keeping residues Ser(168,170) of NFATc4, to regulate subcellular distribution. These data also expand the repertoire of physiological substrates of mTOR and ERK5.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , NFATC Transcription Factors/metabolism , Protein Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Antibodies, Monoclonal , Antibody Specificity , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 7/deficiency , Mitogen-Activated Protein Kinase 7/genetics , Models, Biological , NFATC Transcription Factors/chemistry , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , TOR Serine-Threonine Kinases , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Hybridoma (Larchmt) ; 26(6): 407-15, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18158786

ABSTRACT

To better understand the mechanisms that regulate p120-catenin (p120) and E-cadherin function, we are systematically generating phospho-specific monoclonal antibodies (MAb) to the major p120 phosphorylation sites. p120 has emerged recently as a master regulator of E-cadherin stability and an important modulator of RhoGTPase activities. A number of phosphorylation sites have been identified, but none have as yet been linked to specific regulatory roles. Here, we describe a novel phospho-specific monoclonal antibody to the major PKC-induced p120 phosphorylation site, phospho-serine 879 (pS879). With a few exceptions, p120 MAb pS879 is remarkably specific for the phosphorylated S879 epitope and works effectively in common applications such as Western blot analysis, immunoprecipitation, and immunofluorescence. p120 MAb pS879 should facilitate efforts to identify the role of S879 phosphorylation and to map signaling pathways that modify p120 function through activation of PKC.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Phospho-Specific/biosynthesis , Antibodies, Phospho-Specific/chemistry , Antibody Specificity , Cell Adhesion Molecules/immunology , Phosphoproteins/immunology , Serine/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Phospho-Specific/metabolism , COS Cells , Catenins , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Dogs , Epitopes/immunology , HCT116 Cells , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Rats , Serine/genetics , Serine/metabolism , Delta Catenin
6.
J Exp Med ; 204(1): 79-91, 2007 Jan 22.
Article in English | MEDLINE | ID: mdl-17190839

ABSTRACT

The molecular events involved in the establishment and maintenance of CD4+ central memory and effector memory T cells (TCM and TEM, respectively) are poorly understood. In this study, we demonstrate that ex vivo isolated TCM are more resistant to both spontaneous and Fas-induced apoptosis than TEM and have an increased capacity to proliferate and persist in vitro. Using global gene expression profiling, single cell proteomics, and functional assays, we show that the survival of CD4+ TCM depends, at least in part, on the activation and phosphorylation of signal transducer and activator of transcription 5a (STAT5a) and forkhead box O3a (FOXO3a). TCM showed a significant increase in the levels of phosphorylation of STAT5a compared with TEM in response to both IL-2 (P<0.04) and IL-7 (P<0.002); the latter is well known for its capacity to enhance T cell survival. Moreover, ex vivo TCM express higher levels of the transcriptionally inactive phosphorylated forms of FOXO3a and concomitantly lower levels of the proapoptotic FOXO3a target, Bim. Experiments aimed at blocking FOXO3a phosphorylation confirmed the role of this phosphoprotein in protecting TCM from apoptosis. Our results provide, for the first time in humans, an insight into molecular mechanisms that could be responsible for the longevity and persistence of CD4+ TCM.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Dendritic Cells/immunology , Forkhead Box Protein O3 , Gene Expression Profiling , Humans , I-kappa B Kinase/antagonists & inhibitors , Immunologic Memory , In Vitro Techniques , Lymphocyte Activation , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Suppressor Proteins , fas Receptor/metabolism
7.
J Biomol Screen ; 11(3): 236-46, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16490771

ABSTRACT

A method for high-throughput cloning and analysis of short hairpin RNAs (shRNAs) is described. Using this approach, 464 shRNAs against 116 different genes were screened for knockdown efficacy, enabling rapid identification of effective shRNAs against 74 genes. Statistical analysis of the effects of various criteria on the activity of the shRNAs confirmed that some of the rules thought to govern small interfering RNA (siRNA) activity also apply to shRNAs. These include moderate GC content, absence of internal hairpins, and asymmetric thermal stability. However, the authors did not find strong support for position specific rules. In addition, analysis of the data suggests that not all genes are equally susceptible to RNA interference (RNAi).


Subject(s)
RNA Interference , RNA/chemistry , Base Sequence , Blotting, Western , Cell Line , Cloning, Molecular , DNA Primers , Humans , Polymerase Chain Reaction
8.
Biochemistry ; 43(43): 13694-706, 2004 Nov 02.
Article in English | MEDLINE | ID: mdl-15504032

ABSTRACT

Caveolin-2 is an accessory molecule and the binding partner of caveolin-1. Previously, we showed that c-Src expression leads to the tyrosine phosphorylation of Cav-2 at position 19. To further investigate the tyrosine phosphorylation of Cav-2, we have now generated a novel phospho-specific antibody directed against phospho-Cav-2 (pY27). Here, we show that Cav-2 is phosphorylated at both tyrosines 19 and 27. We reconstituted this phosphorylation event by recombinantly coexpressing c-Src and Cav-2. We generated a series of Cav-2 constructs harboring the mutation of each tyrosine to alanine, singly or in combination, i.e., Cav-2 Y19A, Y27A, and Y19A/Y27A. Recombinant expression of these mutants in Cos-7 cells demonstrated that neither tyrosine is the unique phosphorylation site, and that double mutation of tyrosines 19 and 27 to alanine abrogates Cav-2 tyrosine phosphorylation. Immunofluorescence analysis of NIH 3T3 cells revealed that the two tyrosine-phosphorylated forms of Cav-2 exhibited some distinct properties. Phospho-Cav-2 (pY19) is concentrated at cell edges and at cell-cell contacts, whereas phospho-Cav-2 (pY27) is distributed in a dotlike pattern throughout the cell surface and cytoplasm. Further functional analysis revealed that tyrosine phosphorylation of Cav-2 has no effect on its targeting to lipid rafts, but clearly disrupts the hetero-oligomerization of Cav-2 with Cav-1. In an attempt to identify upstream mediators, we investigated Cav-2 tyrosine phosphorylation in an endogenous setting. We found that in A431 cells, EGF stimulation is sufficient to induce Cav-2 phosphorylation at tyrosines 19 and 27. However, the behavior of the two phosphorylated forms of Cav-2 diverges upon EGF stimulation. First, phospho-Cav-2 (pY19) and phospho-Cav-2 (pY27) display different localization patterns. In addition, the temporal response to EGF stimulation appears to be different. Cav-2 is phosphorylated at tyrosine 19 in a rapid and transient fashion, whereas phosphorylation at tyrosine 27 is sustained over time. Three SH2 domain-containing proteins, c-Src, Nck, and Ras-GAP, were found to associate with Cav-2 in a phosphorylation-dependent manner. However, phosphorylation at tyrosine 27 appears to be more critical than phosphorylation at tyrosine 19 for this binding to occur. Taken together, these results suggest that, in addition to the common characteristics that these two sites appear to share, phospho-Cav-2 (pY19) and phospho-Cav-2 (pY27) may each possess a set of unique functional roles.


Subject(s)
Caveolins/metabolism , Phosphotyrosine/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Substitution/genetics , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Antibody Specificity , Binding Sites, Antibody , COS Cells , Caveolin 2 , Caveolins/genetics , Caveolins/immunology , Cell Line, Transformed , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Humans , Membrane Microdomains/metabolism , Mice , Molecular Weight , NIH 3T3 Cells , Oncogene Proteins/metabolism , Phosphorylation , Phosphotyrosine/genetics , Protein Binding , Proto-Oncogene Proteins pp60(c-src)/metabolism , Proto-Oncogene Proteins pp60(c-src)/physiology , Subcellular Fractions/metabolism , Transfection , ras GTPase-Activating Proteins/metabolism , src Homology Domains
9.
Hybrid Hybridomics ; 23(6): 343-51, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15684660

ABSTRACT

p120-catenin (p120) regulates cadherin turnover and is required for cadherin stability. This role is probably regulated by signaling events that induce p120 phosphorylation, but monitoring individual phosphorylation events and their consequences is technically challenging. Previously, we used phospho-tryptic peptide mapping to identify eight major sites of p120 serine and threonine phosphorylation. Here, we have generated new phospho-specific p120 monoclonal and polyclonal antibodies to phospho-epitopes containing S268, S288, T310, and T910. We have characterized the antibodies with respect to their capabilities and limitations in commonly used assays, including immunoprecipitation (IP), Western blotting (WB), and immunofluorescence (IF). The antibodies should markedly accelerate efforts to delineate the roles of individual p120 modifications and will be particularly useful in identifying upstream signaling events that regulate p120 function.


Subject(s)
Cell Adhesion Molecules/immunology , Phosphoproteins/immunology , Serine/immunology , Threonine/immunology , Animals , COS Cells , Catenins , Chlorocebus aethiops , Dogs , Fluorescent Antibody Technique , Humans , Mice , Phosphorylation , Species Specificity , Delta Catenin
10.
Biochemistry ; 42(23): 7110-23, 2003 Jun 17.
Article in English | MEDLINE | ID: mdl-12795607

ABSTRACT

beta-Dystroglycan is a ubiquitously expressed integral membrane protein that undergoes tyrosine phosphorylation in an adhesion-dependent manner. Tyrosine 892 is now thought to be the principal site for recognition by the c-Src tyrosine kinase; however, little is known about the regulation of this phosphorylation event in vivo. Here, we generated a novel monoclonal antibody probe that recognizes only tyrosine 892 phosphorylated beta-dystroglycan (pY892). We show that upon tyrosine phosphorylation, beta-dystroglycan undergoes a profound change in its sub-cellular localization (e.g., from the plasma membrane to an internal membrane compartment). One possibility is that the net negative charge at position 892 causes the redistribution of beta-dystroglycan to this intracellular vesicular location. In support of this notion, mutation of tyrosine 892 to glutamate (Y892E) is sufficient to drive this intracellular localization, while other point mutants (Y892F and Y892A) remain at the plasma membrane. Interestingly, our colocalization studies with endosomal markers (EEA1, transferrin, and transferrin receptor) suggest that these phospho-beta-dystroglycan containing internal vesicles represent a subset of recycling endosomes. At the level of these internal vesicular structures, we find that tyrosine phosphorylated beta-dystroglycan is colocalized with c-Src. In addition, we demonstrate that known ligands for alpha-dystroglycan, namely, agrin and laminin, are able to induce the tyrosine phosphorylation of beta-dystroglycan. Finally, we show that tyrosine phosphorylated beta-dystroglycan is also detectable in skeletal muscle tissue lysates and is localized to an internal vesicular membrane compartment in skeletal muscle fibers in vivo. The generation of a phospho-specific beta-dystroglycan (pY892) mAb probe provides a new powerful tool for dissecting the role of dystroglycan phosphorylation in normal cellular functioning and in the pathogenesis of muscular dystrophies.


Subject(s)
Cytoskeletal Proteins/metabolism , Intracellular Membranes/metabolism , Membrane Glycoproteins/metabolism , Muscle, Skeletal/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal/chemistry , Biomarkers , COS Cells , Cell Compartmentation/physiology , Cytoskeletal Proteins/genetics , Dystroglycans , Endosomes/chemistry , Endosomes/ultrastructure , Humans , Membrane Glycoproteins/genetics , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/chemistry , Proto-Oncogene Proteins pp60(c-src)/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...